• 沒有找到結果。

In conclusion, we provide for the first time that gefitinib can induce cancer cell apoptosis through an EGFR-independent pathway. Understanding the mechanisms by which securin and ATF3 signal transduction pathways regulate apoptosis following treatment with gefitinib may contribute to the novel therapeutic strategies in cancers.

References

[1] A.J. Levine, The tumor suppressor genes. Annu Rev Biochem 62 (1993) 623-651.

[2] T. de La Motte Rouge, M.C. Petrella, J. Michels, C. Even, C. Balleyguier, J. Duclos, R. Mazeron, P. Morice, P. Pautier, C. Lhomme, New drugs and targeted therapeutic agents in ovarian cancer. Bull Cancer 96 (2009) 1215-1224.

[3] N. Steeghs, J.W. Nortier, H. Gelderblom, Small molecule tyrosine kinase inhibitors in the treatment of solid tumors: an update of recent developments. Ann Surg Oncol 14 (2007) 942-953.

[4] M. Sibilia, R. Kroismayr, B.M. Lichtenberger, A. Natarajan, M. Hecking, M.

Holcmann, The epidermal growth factor receptor: from development to tumorigenesis. Differentiation 75 (2007) 770-787.

[5] A. Citri, Y. Yarden, EGF-ERBB signalling: towards the systems level. Nat Rev Mol Cell Biol 7 (2006) 505-516.

[6] D.J. Riese, E.D. Kim, K. Elenius, S. Buckley, M. Klagsbrun, G.D. Plowman, D.F.

Stern, The epidermal growth factor receptor couples transforming growth factor-alpha, heparin-binding epidermal growth factor-like factor, and amphiregulin to Neu, ErbB-3, and ErbB-4. J Biol Chem 271 (1996) 20047-20052.

[7] M.F. Press, H.J. Lenz, EGFR, HER2 and VEGF pathways: validated targets for cancer treatment. Drugs 67 (2007) 2045-2075.

[8] R.S. Herbst, J.V. Heymach, S.M. Lippman, Lung cancer. N Engl J Med 359 (2008) 1367-1380.

[9] F. Ciardiello, G. Tortora, EGFR antagonists in cancer treatment. N Engl J Med 358 (2008) 1160-1174.

[10] C. Ho, J. Laskin, EGFR-directed therapies to treat non-small-cell lung cancer.

Expert Opin Investig Drugs 18 (2009) 1133-1145.

[11] S.A. Aaronson, Growth factors and cancer. Science 254 (1991) 1146-1153.

[12] J.R. Grandis, J.C. Sok, Signaling through the epidermal growth factor receptor during the development of malignancy. Pharmacol Ther 102 (2004) 37-46.

[13] A.W. Hemming, N.L. Davis, A. Kluftinger, B. Robinson, N.F. Quenville, B.

Liseman, J. LeRiche, Prognostic markers of colorectal cancer: an evaluation of DNA content, epidermal growth factor receptor, and Ki-67. J Surg Oncol 51 (1992) 147-152.

[14] P.R. Dutta, A. Maity, Cellular responses to EGFR inhibitors and their relevance to cancer therapy. Cancer Lett 254 (2007) 165-177.

[15] P.M. Harari, Epidermal growth factor receptor inhibition strategies in oncology.

Endocr Relat Cancer 11 (2004) 689-708.

[16] J. Baselga, Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist 7 Suppl 4 (2002) 2-8.

[17] J. Baselga, S.D. Averbuch, ZD1839 ('Iressa') as an anticancer agent. Drugs 60 Suppl 1 (2000) 33-40; discussion 41-32.

[18] G. Lurje, H.J. Lenz, EGFR signaling and drug discovery. Oncology 77 (2009) 400- 410.

[19] T. Takano, Y. Ohe, M. Kusumoto, U. Tateishi, S. Yamamoto, H. Nokihara, N.

Yamamoto, I. Sekine, H. Kunitoh, T. Tamura, T. Kodama, N. Saijo, Risk factors for interstitial lung disease and predictive factors for tumor response in patients with advanced non-small cell lung cancer treated with gefitinib. Lung Cancer 45 (2004) 93-104.

[20] F. Ciardiello, R. Caputo, R. Bianco, V. Damiano, G. Pomatico, S. De Placido, A.R.

Bianco, G. Tortora, Antitumor effect and potentiation of cytotoxic drugs activity in human cancer cells by ZD-1839 (Iressa), an epidermal growth factor receptor- selective tyrosine kinase inhibitor. Clin Cancer Res 6 (2000) 2053-2063.

[21] J.G. Paez, P.A. Janne, J.C. Lee, S. Tracy, H. Greulich, S. Gabriel, P. Herman, F.J.

Kaye, N. Lindeman, T.J. Boggon, K. Naoki, H. Sasaki, Y. Fujii, M.J. Eck, W.R.

Sellers, B.E. Johnson, M. Meyerson, EGFR mutations in lung cancer:

correlation with clinical response to gefitinib therapy. Science 304 (2004) 1497- 1500.

[22] M. Ono, A. Hirata, T. Kometani, M. Miyagawa, S. Ueda, H. Kinoshita, T. Fujii, M.

Kuwano, Sensitivity to gefitinib (Iressa, ZD1839) in non-small cell lung cancer cell lines correlates with dependence on the epidermal growth factor (EGF) receptor/extracellular signal-regulated kinase 1/2 and EGF receptor/Akt pathway

for proliferation. Mol Cancer Ther 3 (2004) 465-472.

[23] F.M. Sirotnak, M.F. Zakowski, V.A. Miller, H.I. Scher, M.G. Kris, Efficacy of cytotoxic agents against human tumor xenografts is markedly enhanced by coadministration of ZD1839 (Iressa), an inhibitor of EGFR tyrosine kinase. Clin Cancer Res 6 (2000) 4885-4892.

[24] I. Okamoto, Epidermal growth factor receptor in relation to tumor development:

EGFR-targeted anticancer therapy. FEBS J 277 (2010) 309-315.

[25] E.R. Camp, J. Summy, T.W. Bauer, W. Liu, G.E. Gallick, L.M. Ellis, Molecular mechanisms of resistance to therapies targeting the epidermal growth factor receptor. Clin Cancer Res 11 (2005) 397-405.

[26] M.D. Jacobson, M. Weil, M.C. Raff, Programmed cell death in animal development. Cell 88 (1997) 347-354.

[27] M.C. Raff, Social controls on cell survival and cell death. Nature 356 (1992) 397- 400.

[28] M.C. Raff, B.A. Barres, J.F. Burne, H.S. Coles, Y. Ishizaki, M.D. Jacobson, Programmed cell death and the control of cell survival. Philos Trans R Soc Lond B Biol Sci 345 (1994) 265-268.

[29] J.F. Kerr, A.H. Wyllie, A.R. Currie, Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26 (1972) 239- 257.

[30] D.R. Green, G. Kroemer, The pathophysiology of mitochondrial cell death. Science 305 (2004) 626-629.

[31] S.J. Martin, D.R. Green, Protease activation during apoptosis: death by a thousand cuts? Cell 82 (1995) 349-352.

[32] T. Patel, G.J. Gores, S.H. Kaufmann, The role of proteases during apoptosis.

FASEB J 10 (1996) 587-597.

[33] M. Weil, M.D. Jacobson, H.S. Coles, T.J. Davies, R.L. Gardner, K.D. Raff, M.C.

Raff, Constitutive expression of the machinery for programmed cell death. J Cell Biol 133 (1996) 1053-1059.

[34] A.M. Chinnaiyan, W.L. Hanna, K. Orth, H. Duan, G.G. Poirier, C.J. Froelich, V.M.

Dixit, Cytotoxic T-cell-derived granzyme B activates the apoptotic protease ICE-LAP3. Curr Biol 6 (1996) 897-899.

[35] E.S. Alnemri, Mammalian cell death proteases: a family of highly conserved aspartate specific cysteine proteases. J Cell Biochem 64 (1997) 33-42.

[36] L.M. Martins, T. Kottke, P.W. Mesner, G.S. Basi, S. Sinha, N. Frigon, Jr., E. Tatar, J.S. Tung, K. Bryant, A. Takahashi, P.A. Svingen, B.J. Madden, D.J.

McCormick, W.C. Earnshaw, S.H. Kaufmann, Activation of multiple interleukin-1beta converting enzyme homologues in cytosol and nuclei of HL-60 cells during etoposide-induced apoptosis. J Biol Chem 272 (1997) 7421-7430.

[37] P.J. Duriez, G.M. Shah, Cleavage of poly(ADP-ribose) polymerase: a sensitive parameter to study cell death. Biochem Cell Biol 75 (1997) 337-349.

[38] D.K. Miller, The role of the Caspase family of cysteine proteases in apoptosis.

Semin Immunol 9 (1997) 35-49.

[39] C.B. Thompson, Apoptosis in the pathogenesis and treatment of disease. Science 267 (1995) 1456-1462.

[40] K. Vermeulen, D.R. Van Bockstaele, Z.N. Berneman, Apoptosis: mechanisms and relevance in cancer. Ann Hematol 84 (2005) 627-639.

[41] K. Labedzka, A. Grzanka, M. Izdebska, [Mitochondria and cell death]. Postepy Hig Med Dosw (Online) 60 (2006) 439-446.

[42] S.J. Hong, T.M. Dawson, V.L. Dawson, Nuclear and mitochondrial conversations in cell death: PARP-1 and AIF signaling. Trends Pharmacol Sci 25 (2004) 259- 264.

[43] T. Eisenberg, S. Buttner, G. Kroemer, F. Madeo, The mitochondrial pathway in yeast apoptosis. Apoptosis 12 (2007) 1011-1023.

[44] L. Pei, S. Melmed, Isolation and characterization of a pituitary tumor-transforming gene (PTTG). Mol Endocrinol 11 (1997) 433-441.

[45] H. Funabiki, H. Yamano, K. Kumada, K. Nagao, T. Hunt, M. Yanagida, Cut2 proteolysis required for sister-chromatid seperation in fission yeast. Nature 381 (1996) 438-441.

[46] A. Yamamoto, V. Guacci, D. Koshland, Pds1p, an inhibitor of anaphase in budding yeast, plays a critical role in the APC and checkpoint pathway(s). J Cell Biol 133 (1996) 99-110.

[47] H. Zou, T.J. McGarry, T. Bernal, M.W. Kirschner, Identification of a vertebrate sister-chromatid separation inhibitor involved in transformation and

tumorigenesis. Science 285 (1999) 418-422.

[48] A. Dominguez, F. Ramos-Morales, F. Romero, R.M. Rios, F. Dreyfus, M.

Tortolero, J.A. Pintor-Toro, hpttg, a human homologue of rat pttg, is overexpressed in hematopoietic neoplasms. Evidence for a transcriptional activation function of hPTTG. Oncogene 17 (1998) 2187-2193.

[49] K. Nasmyth, Disseminating the genome: joining, resolving, and separating sister chromatids during mitosis and meiosis. Annu Rev Genet 35 (2001) 673-745.

[50] O. Stemmann, H. Zou, S.A. Gerber, S.P. Gygi, M.W. Kirschner, Dual inhibition of sister chromatid separation at metaphase. Cell 107 (2001) 715-726.

[51] P.V. Jallepalli, I.C. Waizenegger, F. Bunz, S. Langer, M.R. Speicher, J.M. Peters, K.W. Kinzler, B. Vogelstein, C. Lengauer, Securin is required for chromosomal stability in human cells. Cell 105 (2001) 445-457.

[52] K. Nagao, Y. Adachi, M. Yanagida, Separase-mediated cleavage of cohesin at interphase is required for DNA repair. Nature 430 (2004) 1044-1048.

[53] X. Zhang, G.A. Horwitz, A.P. Heaney, M. Nakashima, T.R. Prezant, M.D.

Bronstein, S. Melmed, Pituitary tumor transforming gene (PTTG) expression in pituitary adenomas. J Clin Endocrinol Metab 84 (1999) 761-767.

[54] C. Solbach, M. Roller, C. Fellbaum, M. Nicoletti, M. Kaufmann, PTTG mRNA expression in primary breast cancer: a prognostic marker for lymph node invasion and tumor recurrence. Breast 13 (2004) 80-81.

[55] R. Puri, A. Tousson, L. Chen, S.S. Kakar, Molecular cloning of pituitary tumor transforming gene 1 from ovarian tumors and its expression in tumors. Cancer Lett 163 (2001) 131-139.

[56] S.S. Kakar, M.T. Malik, Suppression of lung cancer with siRNA targeting PTTG.

Int J Oncol 29 (2006) 387-395.

[57] A.P. Heaney, R. Singson, C.J. McCabe, V. Nelson, M. Nakashima, S. Melmed, Expression of pituitary-tumour transforming gene in colorectal tumours. Lancet 355 (2000) 716-719.

[58] X. Zhang, G.A. Horwitz, T.R. Prezant, A. Valentini, M. Nakashima, M.D.

Bronstein, S. Melmed, Structure, expression, and function of human pituitary tumor-transforming gene (PTTG). Mol Endocrinol 13 (1999) 156-166.

[59] T. Hamid, M.T. Malik, S.S. Kakar, Ectopic expression of PTTG1/securin promotes

tumorigenesis in human embryonic kidney cells. Mol Cancer 4 (2005) 3.

[60] K. Boelaert, C.J. McCabe, L.A. Tannahill, N.J. Gittoes, R.L. Holder, J.C.

Watkinson, A.R. Bradwell, M.C. Sheppard, J.A. Franklyn, Pituitary tumor transforming gene and fibroblast growth factor-2 expression: potential prognostic indicators in differentiated thyroid cancer. J Clin Endocrinol Metab 88 (2003) 2341-2347.

[61] G. Liang, C.D. Wolfgang, B.P. Chen, T.H. Chen, T. Hai, ATF3 gene. Genomic organization, promoter, and regulation. J Biol Chem 271 (1996) 1695-1701.

[62] T. Hai, M.G. Hartman, The molecular biology and nomenclature of the activating transcription factor/cAMP responsive element binding family of transcription factors: activating transcription factor proteins and homeostasis. Gene 273 (2001) 1-11.

[63] D. Lu, J. Chen, T. Hai, The regulation of ATF3 gene expression by mitogen- activated protein kinases. Biochem J 401 (2007) 559-567.

[64] M.R. Montminy, L.M. Bilezikjian, Binding of a nuclear protein to the cyclic-AMP response element of the somatostatin gene. Nature 328 (1987) 175-178.

[65] P.J. Deutsch, J.P. Hoeffler, J.L. Jameson, J.C. Lin, J.F. Habener, Structural determinants for transcriptional activation by cAMP-responsive DNA elements.

J Biol Chem 263 (1988) 18466-18472.

[66] B.P. Chen, C.D. Wolfgang, T. Hai, Analysis of ATF3, a transcription factor induced by physiological stresses and modulated by gadd153/Chop10. Mol Cell Biol 16 (1996) 1157-1168.

[67] M.G. Hartman, D. Lu, M.L. Kim, G.J. Kociba, T. Shukri, J. Buteau, X. Wang, W.L.

Frankel, D. Guttridge, M. Prentki, S.T. Grey, D. Ron, T. Hai, Role for activating transcription factor 3 in stress-induced beta-cell apoptosis. Mol Cell Biol 24 (2004) 5721-5732.

[68] M.R. Thompson, D. Xu, B.R. Williams, ATF3 transcription factor and its emerging roles in immunity and cancer. J Mol Med 87 (2009) 1053-1060.

[69] X. Yin, J.W. Dewille, T. Hai, A potential dichotomous role of ATF3, an adaptive- response gene, in cancer development. Oncogene 27 (2008) 2118-2127.

[70] A.E. Pelzer, J. Bektic, P. Haag, A.P. Berger, A. Pycha, G. Schafer, H. Rogatsch, W.

Horninger, G. Bartsch, H. Klocker, The expression of transcription factor activating transcription factor 3 in the human prostate and its regulation by androgen in prostate cancer. J Urol 175 (2006) 1517-1522.

[71] S. Bandyopadhyay, Y. Wang, R. Zhan, S.K. Pai, M. Watabe, M. Iiizumi, E. Furuta, S. Mohinta, W. Liu, S. Hirota, S. Hosobe, T. Tsukada, K. Miura, Y. Takano, K.

Saito, T. Commes, D. Piquemal, T. Hai, K. Watabe, The tumor metastasis suppressor gene Drg-1 down-regulates the expression of activating transcription factor 3 in prostate cancer. Cancer Res 66 (2006) 11983-11990.

[72] X. Huang, X. Li, B. Guo, KLF6 induces apoptosis in prostate cancer cells through up-regulation of ATF3. J Biol Chem 283 (2008) 29795-29801.

[73] F.G. Bottone, Jr., Y. Moon, J.S. Kim, B. Alston-Mills, M. Ishibashi, T.E. Eling, The anti-invasive activity of cyclooxygenase inhibitors is regulated by the transcription factor ATF3 (activating transcription factor 3). Mol Cancer Ther 4 (2005) 693-703.

[74] F. Ciardiello, G. Tortora, A novel approach in the treatment of cancer: targeting the epidermal growth factor receptor. Clin Cancer Res 7 (2001) 2958-2970.

[75] S.K. Grant, Therapeutic protein kinase inhibitors. Cell Mol Life Sci 66 (2009) 1163-1177.

[76] A. Arora, E.M. Scholar, Role of tyrosine kinase inhibitors in cancer therapy. J Pharmacol Exp Ther 315 (2005) 971-979.

[77] P. Meier, A. Finch, G. Evan, Apoptosis in development. Nature 407 (2000) 796- 801.

[78] Y. Shi, Mechanisms of caspase activation and inhibition during apoptosis. Mol Cell 9 (2002) 459-470.

[79] Q.A. Liu, M.O. Hengartner, The molecular mechanism of programmed cell death in C. elegans. Ann N Y Acad Sci 887 (1999) 92-104.

[80] A. Degterev, M. Boyce, J. Yuan, A decade of caspases. Oncogene 22 (2003) 8543- 8567.

[81] F. Romero, A.M. Gil-Bernabe, C. Saez, M.A. Japon, J.A. Pintor-Toro, M.

Tortolero, Securin is a target of the UV response pathway in mammalian cells.

Mol Cell Biol 24 (2004) 2720-2733.

[82] Y. Zhou, K.R. Mehta, A.P. Choi, S. Scolavino, X. Zhang, DNA damage-induced inhibition of securin expression is mediated by p53. J Biol Chem 278 (2003) 462-470.

[83] S.J. Chiu, T.S. Hsu, J.I. Chao, Opposing securin and p53 protein expression in the oxaliplatin-induced cytotoxicity of human colorectal cancer cells. Toxicol Lett 167 (2006) 122-130.

[84] S.J. Chiu, J.I. Chao, Y.J. Lee, T.S. Hsu, Regulation of gamma-H2AX and securin contribute to apoptosis by oxaliplatin via a p38 mitogen-activated protein kinase-dependent pathway in human colorectal cancer cells. Toxicol Lett 179 (2008) 63-70.

[85] J.I. Chao, H.F. Liu, The blockage of survivin and securin expression increases the cytochalasin B-induced cell death and growth inhibition in human cancer cells.

Mol Pharmacol 69 (2006) 154-164.

[86] J.I. Chao, S.H. Hsu, T.C. Tsou, Depletion of securin increases arsenite-induced chromosome instability and apoptosis via a p53-independent pathway. Toxicol Sci 90 (2006) 73-86.

[87] T.W. Hai, F. Liu, W.J. Coukos, M.R. Green, Transcription factor ATF cDNA clones: an extensive family of leucine zipper proteins able to selectively form DNA-binding heterodimers. Genes Dev 3 (1989) 2083-2090.

[88] T. Hai, T. Curran, Cross-family dimerization of transcription factors Fos/Jun and ATF/CREB alters DNA binding specificity. Proc Natl Acad Sci U S A 88 (1991) 3720-3724.

[89] J.C. Hsu, R. Bravo, R. Taub, Interactions among LRF-1, JunB, c-Jun, and c-Fos define a regulatory program in the G1 phase of liver regeneration. Mol Cell Biol 12 (1992) 4654-4665.

[90] T. Hai, C.D. Wolfgang, D.K. Marsee, A.E. Allen, U. Sivaprasad, ATF3 and stress responses. Gene Expr 7 (1999) 321-335.

[91] D. Lu, C.D. Wolfgang, T. Hai, Activating transcription factor 3, a stress-inducible gene, suppresses Ras-stimulated tumorigenesis. J Biol Chem 281 (2006) 10473- 10481.

[92] Y. Kang, C.R. Chen, J. Massague, A self-enabling TGFbeta response coupled to stress signaling: Smad engages stress response factor ATF3 for Id1 repression in

epithelial cells. Mol Cell 11 (2003) 915-926.

[93] M.T. Ling, X. Wang, X. Zhang, Y.C. Wong, The multiple roles of Id-1 in cancer progression. Differentiation 74 (2006) 481-487.

Gefitinib (μM, 24 h)

Fig. 1. The effect of gefitinib on the cell viability in a variety of human cancer cell lines. The cells were treated with 0-60 μM gefitinib for 24 h. After drug

treatment, the cells were washed with PBS and incubated for 2 days. The cell viability was measured by MTT assay. The results were obtained from four to seven experiments and the bar represents the mean ± S.E.M. *p < 0.05 and **p

< 0.01 indicate significant difference between control and gefitinib treated samples.

Fig. 2. The protein level of phospho-EGFR and total EGFR in a variety of human cancer cell lines. The total protein extracts from various cancer cell lines including RKO, A549, BFTC905, MCF7 and A375 were subjected to western blot analysis using specific anti-phospho-EGFR, anti-EGFR and anti-actin antibodies. The representative western blot data were shown from one of three separate experiments with similar findings. Actin was a loading control. Positive

control of phospo-EGFR and EGFR were extracted proteins from A431 cells.

(A)

Fig. 3. The effect of gefitinib on the cell cycle progression and sub-G1 formation in A375 and BFTC905 cells. (A) The cells were treated with 0-60 μM gefitinib for

(A)

Fig. 4. The effect of gefitinib on apoptosis in A375 and BFTC905 cells. (A) After .treatment with or without gefitinib for 24 h, the cells collected and then .subjected to annexin V and PI staining. The representative flow data were .shown from one of three to four separate experiments with similar findings. The .results represented the mean ± S.E.M. (B) The percentage of apoptosis .populations (early and late stages) were quantified by CellQuest software. The .results were obtained from three to four experiments and the bar represents the .mean ± S.E.M. **p < 0.01 indicates significant difference between control and

Annexin V integral

Fig. 5. The time-lapse observation of apoptosis by gefitinib. A375 cells were plated at a density of 2 × 105 cells/p35 Petri dish for 24 h, then the cells were treated with or without 60 μM gefitinib for 24 h. The cell morphology was observed under a living cell imaging system. The representative cell morphology data were shown from one of three separate experiments with similar findings. The

arrows indicated the apoptotic cells.

0 h 8 h 16 h 24 h

A375 cells (gefitinib 60 μM)

0 h 8 h 16 h 24 h

A375 cells (control)

10 μm

10 μm

(A)

(B)

A375 cells

Gefitinib (μM, 24 h)

0 20 40

Mitochondrial membrance potential (intensity of DiOC6)

0

Fig. 6. The effect of gefitinib on the hyperpolarization of mitochondria. (A) After .treatment with or without gefitinib for 24 h, the cells were incubated with 50 .nM DiOC6 and then analyzed by a flow cytometer. The representative flow .data were shown from one of five separate experiments with similar findings.

.(B) The fluorescence intensity of DiOC6 were quantified by CellQuest software.

.The results were obtained from five experiments and the bar represents the .mean ± S.E.M. *p < 0.05 and **p < 0.01 indicate significant difference between

Cell counts

Fig. 7. The effect of gefitinib on the activation of caspase-3 and the cleavage of

(A)

(B)

A375 cells

Gefitinib (μM, 24 h)

0 10 20 40 60

Relative protein level of securin (fold) 0.2

0.4

Fig. 8. The effect of gefitinib on the securin protein expression. (A) A375 cells were treated with 0-60 μM gefitinib for 24 h. The total protein extracts were prepared for immunoblot analysis using anti-securin and anti-actin antibodies. The representative western blot data were shown from one of three separate experiments with similar findings. Actin was a loading control. (B) The relative protein level of securin were from western blot by semi-quantification. The results were obtained from three experiments and the bar represents the mean ± S.E.M. *p < 0.05 and **p < 0.01 indicate significant difference between control and gefitinib treated samples.

Fig. 9. The effect of gefitinib on the expression and location of securin. A375 cells were treated with or without 40 μM gefitinib for 24 h. At the end of treatment, the cells were incubated with mouse anti-securin antibody and then incubated with goat anti-mouse FITC. The β-tubulin and nuclei were stained with the Cy3- labeled anti-β-tubulin and Hoechst 33258, respectively.

control

gefitinib (40 μM, 24 h)

control

gefitinib (40 μM, 24 h)

nuclei β-tubulin securin merge

A375 cells

100 μm

100 μm

20 μm

20 μm

HCT116 cells

Gefitinib (μM, 24 h)

0 10 20 40 60

Cell viabilit y ( % )

10 20 30 40 50 6070 8090 100110 120

securin (+/+) securin (-/-)

**

**

** **

* *

#

Fig. 10. The comparison of cytotoxicity between the securin-wild type and -null .HCT116 cancer cells following gefitinib treatment. The cells were treated .with 0-60 μM gefitinib for 24 h. After drug treatment, the cells were washed .with PBS and incubated for 2 days. The cell viability was measured by MTT .assay. The results were obtained from six experiments and the bar represents .the mean ± S.E.M. *p < 0.05 and **p < 0.01 indicate significant difference .between control and gefitinib treated samples. #p < 0.05 indicates significant .difference between the securin-wild type and -null HCT116 cancer cells by the .gefitinib treatment at the same concentration.

A375 cells

Fig. 11. The effect of the overexpression of securin on the gefitinib-induced cytotoxicity. The cells were transfected the control or securin vectors and

(A)

(B)

A375 cells

Gefitinib (μM, 24 h)

0 10 20 40 60

Relative protein level of ATF3 (fold) 0.5

1.0 1.5 2.0 2.5 3.0

*

**

Fig. 12. The effect of gefitinib on the ATF3 protein expression. (A) A375 cells were treated with 0-60 μM gefitinib for 24 h. The total protein extracts were prepared for immunoblot analysis using anti-ATF3 and anti-actin antibodies.

The representative western blot data were shown from one of three separate experiments with similar findings. Actin was a loading control. (B) The relative protein level of ATF3 were from western blot by semi-quantification.

The results were obtained from three experiments and the bar represents the mean ± S.E.M. *p < 0.05 and **p < 0.01 indicate significant difference

A375 cells Gefitinib

(μM, 24 h)

Actin ATF3

0 10 20 40 60

Fig. 13. The effect of gefitinib on the expression and location of ATF3. A375 cells were treated with or without 40 μM gefitinib for 24 h. At the end of treatment, the cells were incubated with rabbit anti-ATF3 antibody and then incubated with goat anti-rabbit Cy-3. The F-actin and nuclei were stained with BODIPY FL phallacidin and Hoechst 33258, respectively.

control

gefitinib (40 μM, 24 h)

control

gefitinib (40 μM, 24 h)

nuclei F-actin ATF3 merge

A375 cells

100 μm

100 μm

20 μm

20 μm

RKO cells

Fig. 14. The comparison of cytotoxicity between the gefitinib and its analogues in a variety of human cancer cell lines. The cells were treated with 0-80 μM gefitinib or its analogues for 24 h. After drug treatment, the cells were washed with PBS and incubated for 2 days. The cell viability was measured by MTT assay. The results were obtained from three to seven experiments and the bar represents the mean ± S.E.M. **p < 0.01 indicates significant difference

(A)

(B)

Fig. 15. The schematic diagram of antitumor activity by gefitinib

Fig. 16. A proposed model for the gefitinib-induced apoptosis

Appendix 1. The structure of gefitinib

相關文件